Elsevier

Immunobiology

Volume 217, Issue 7, July 2012, Pages 676-683
Immunobiology

An anti-IgE monoclonal antibody that binds to IgE on CD23 but not on high-affinity IgE.Fc receptors

https://doi.org/10.1016/j.imbio.2011.11.006Get rights and content

Abstract

A new monoclonal antibody (mAb), specific for human IgE, the central mediator of immediate-type hypersensitivity reactions, has been shown to possess a unique set of binding specificities. The mAb, 8D6, binds to a conformational epitope on the CH3 domain of human e immunoglobulin and can compete with omalizumab for binding to IgE. Like omalizumab, it does not bind to IgE bound by the high-affinity IgE.Fc receptor (FcɛRI) on basophils and mast cells. It also does not cause activation and degranulation of IgE-pulsed, human FcɛRI-expressing rat basophilic leukemic cells (RBL SX-38). The mAb can inhibit IgE binding to recombinant α chain of human FcɛRI in ELISA and to human FcɛRI-expressing RBL SX38 cells in fluorescence flow cytometric analysis. However, unlike omalizumab, 8D6 can bind to IgE already bound by the low-affinity IgE.Fc receptors (FcɛRII, or CD23), as revealed in ELISA with recombinant CD23 and in flow cytometric analysis with human B cells. Since earlier investigators have shown that anti-CD23 mAbs can inhibit the synthesis of IgE in lymphocyte culture in vitro and can down-regulate IgE production in treated patients, 8D6 may offer pharmacological mechanisms in addition to those mediated by omalizumab, for controlling IgE in patients with allergic diseases.

Introduction

Allergic diseases, such as allergic asthma, allergic rhinitis, and food allergy, are very prevalent among populations living a Westernized lifestyle (Gold and Wright, 2005, Isolauri et al., 2004). Numerous clinical studies of the humanized anti-IgE antibody, omalizumab, in many allergic diseases have shown that omalizumab is efficacious and safe in treating patients with those allergic diseases (Lanier et al., 2003, Milgrom et al., 1999, Rafi et al., 2010). These results have not only confirmed that IgE is a central mediator in the pathogenesis of many allergic diseases but have also validated IgE as a good molecular target for therapeutic intervention (Holgate et al. 2005). The widely recognized pharmacologic mechanisms of omalizumab are its ability to neutralize free IgE and to inhibit IgE binding to FcɛRI on basophils and mast cells (Chang, 2000, Chang et al., 2007). Moreover, the depletion of free IgE in the blood and in interstitial spaces causes a down-regulation of FcɛRI on basophils and mast cells over time (Beck et al., 2004, Chang and Shiung, 2006). While the ability of the antibodies to bind to membrane-bound IgE (mIgE) (Davis et al. 1993), which forms part of the B cell receptor complex (Niiro and Clark 2002), and to cause the lysis of IgE-expressing B cells and hence inhibit IgE production, was considered in the original design of the anti-IgE therapeutic antibodies, this property has not been confirmed for omalizumab in patients.

Therapeutic approaches aiming at suppressing IgE production, such as anti-interleukin 4 administration (Hart et al. 2002), TH2/TH1 modulation (Stokes and Casale 2004), and anti-CD23 treatments (Bonnefoy et al., 1990, Poole et al., 2005), have drawn much interest among researchers attempting to develop treatments for IgE-mediated allergic diseases. CD23 expressed on the surface of B cells, which binds to IgE with an association constant Ka of about 108–109 M−1 (Gould and Sutton 2008), is known to be involved in the regulation of IgE production (Conrad et al. 2007). A mature CD23 molecule is formed by three 45-kDa type II transmembrane subunit proteins that belong to the C-type lectin family (Conrad 1990). Each subunit contains an α-helical stalk region between the extracellular lectin domain and the transmembrane domain, which bundles to form coiled-coil trimers (Beavil et al., 1992, Hibbert et al., 2005). Membrane-bound CD23 is cleaved by a metalloproteinase, ADAM10, releasing the secreted form of CD23 (Lemieux et al. 2007). It has been shown in mice in vivo that when the lectin domain of membrane-bound CD23 is bound by IgE or by an anti-CD23 antibody, CD23 becomes more stable and is less susceptible to the cleavage by ADAM 10, which signals the B cell to reduce IgE synthesis (Conrad et al., 2007, Ford et al., 2006). Several chimeric anti-CD23 mAbs were shown to cross-link CD23 and inhibit the transcription of ɛ gene and the production of IgE by human peripheral B cells in culture (Yabuuchi et al. 2002). An anti-CD23 mAb, lumiliximab, was shown to induce apoptosis by cross-linking CD23 on B cell lines or on chronic lymphocytic leukemia cells (Pathan et al. 2008). Furthermore, lumiliximab was able to reduce blood IgE levels in allergic patients (Rosenwasser et al. 2003) and is potentially applicable for treating patients with chronic lymphocytic leukemia (Byrd et al. 2007).

In this study, we have developed an anti-IgE mAb, 8D6, which resembles omalizumab in various aspects, including being able to bind to the same region on the CH3 domain of ɛ chain, not being able to bind to IgE on FcɛRI, and not being able to induce the activation of basophils. However, mAb 8D6, unlike omalizumab, has the additional attribute of being able to bind to IgE that is already bound by CD23 and hence has the potential to down-regulate IgE synthesis.

Section snippets

Cells

Mouse NS0 myeloma cells and hybridoma cells were cultured in DMEM (Invitrogen, Carlsbad, CA) supplemented with 10% heat-inactivated FBS, 4 mM l-glutamine, 50 μg/ml penicillin (Invitrogen), and 100 μg/ml streptomycin (Invitrogen). The RBL SX-38 cell line, provided by Dr. J.P. Kinet of Harvard Medical School, was maintained in MEM (Invitrogen) supplemented with 10% heat-inactivated FBS, 2 mM GlutaMAX (Invitrogen), 25 mM HEPES, 1 mM sodium pyruvate, 50 μg/ml penicillin, and 100 μg/ml streptomycin.

Generation of anti-human IgE mAbs that bind to IgE on FcɛRII but not FcɛRI

To obtain anti-human IgE mAbs with the desired set of binding specificities as described in the introduction, BALB/c mice were immunized with human mIgE.FcL, and hybridoma clones were prepared. In the primary screening procedure, the clones specific for human IgE in the initial ELISA were tested for their ability to compete with omalizumab for binding to IgE. Those that did compete with omalizumab were then screened for their ability to bind to human IgE-pulsed CD23-expressing SKW cells by

Discussion

The anti-IgE therapeutic antibody, omalizumab, has been approved in the United States since 2003 for the treatment of moderate to severe allergic asthma and in the European Union since 2005 for the treatment of severe allergic asthma (Chang et al. 2007). Many patients have been given the same omalizumab dosage continuously for several years without a clearly defined treatment period (Lanier 2006). The general understanding among the clinicians prescribing omalizumab is that IgE is continuously

Acknowledgements

We thank Dr. Bing-Hung Chen for helping us to obtain experimental materials from Dr. D.H. Conrad. This study was supported by a grant, NSC99-2320-B-001-006-MY3, from the National Science Council, Taiwan.

References (35)

  • A.J. Beavil et al.

    Alpha-helical coiled-coil stalks in the low-affinity receptor for IgE (Fc epsilon RII/CD23) and related C-type lectins

    Proc. Natl. Acad. Sci. U.S.A.

    (1992)
  • L.A. Beck et al.

    Omalizumab-induced reductions in mast cell Fc epsilon RI expression and function

    J. Allergy Clin. Immunol.

    (2004)
  • J.Y. Bonnefoy et al.

    Inhibition of human interleukin 4-induced IgE synthesis by a subset of anti-CD23/Fc epsilon RII monoclonal antibodies

    Eur. J. Immunol.

    (1990)
  • J.C. Byrd et al.

    Phase 1 study of lumiliximab with detailed pharmacokinetic and pharmacodynamic measurements in patients with relapsed or refractory chronic lymphocytic leukemia

    Clin. Cancer Res.

    (2007)
  • T.W. Chang

    The pharmacological basis of anti-IgE therapy

    Nat. Biotechnol.

    (2000)
  • T.W. Chang et al.

    Anti-IgE as a mast cell-stabilizing therapeutic agent

    J. Allergy Clin. Immunol.

    (2006)
  • T.W. Chang et al.

    Anti-IgE antibodies for the treatment of IgE-mediated allergic diseases

    Adv. Immunol.

    (2007)
  • J.B. Chen et al.

    Unique epitopes on C epsilon mX in IgE-B cell receptors are potentially applicable for targeting IgE-committed B cells

    J. Immunol.

    (2010)
  • D.H. Conrad

    Fc epsilon RII/CD23: the low affinity receptor for IgE

    Annu. Rev. Immunol.

    (1990)
  • D.H. Conrad et al.

    CD23: an overlooked regulator of allergic disease

    Curr. Allergy Asthma Rep.

    (2007)
  • F.M. Davis et al.

    Can anti-IgE be used to treat allergy?

    Springer Semin. Immunopathol.

    (1993)
  • J.W. Ford et al.

    In vivo murine CD23 destabilization enhances CD23 shedding and IgE synthesis

    Cell. Immunol.

    (2006)
  • D.R. Gold et al.

    Population disparities in asthma

    Annu. Rev. Public Health

    (2005)
  • H.J. Gould et al.

    IgE in allergy and asthma today

    Nat. Rev. Immunol.

    (2008)
  • Q.K. Griffith et al.

    CD23-bound IgE augments and dominates recall responses through human naive B cells

    J. Immunol.

    (2011)
  • T.K. Hart et al.

    Preclinical efficacy and safety of pascolizumab (SB 240683): a humanized anti-interleukin-4 antibody with therapeutic potential in asthma

    Clin. Exp. Immunol.

    (2002)
  • R.G. Hibbert et al.

    The structure of human CD23 and its interactions with IgE and CD21

    J. Exp. Med.

    (2005)
  • Cited by (33)

    • Mucosal IgE immune responses in respiratory diseases

      2019, Current Opinion in Pharmacology
      Citation Excerpt :

      The Fab fragment of the rat anti-IgE Cε2 domain mAb (Fab-6HD5) was shown to antagonize IgE binding to FcεRI in a mouse study [89]. The murine mAb 8D6 binds to human IgE Cε3 domain, but unlike omalizumab, it can also recognize IgE molecules bound to CD23 on the surface of B cells [90]. Designed ankyrin proteins (DARPin) are products of protein engineering, which have been used to antagonize IgE-mediated activation of effector cells [91•].

    • Biologics in the treatment of severe asthma

      2017, Allergologia et Immunopathologia
      Citation Excerpt :

      Currently, omalizumab is the only mAb approved for the treatment of asthma. A new human anti-IgE mAb, 8D6, binds to a conformational epitope on the CH3 domain of human IgE, can bind to already bound IgE to low-affinity receptors (FcɛRII od CD23) and can compete with omalizumab for IgE binding.6 QGE031 (ligelizumab) is an investigational anti-IgE antibody that binds IgE with higher affinity than omalizumab.

    • Allosteric mechanism of action of the therapeutic anti-IgE antibody omalizumab

      2017, Journal of Biological Chemistry
      Citation Excerpt :

      In the FabXol3/IgE-Fc complex, the Cɛ3 domains are positioned further away from one another than in any other crystal structure containing IgE-Fc or Fcɛ3–4 and thus adopt the most open conformation observed thus far (Figs. 1B and 4, C and D, and supplemental Movie S2); this conformation is significantly more open than the conformation for FcɛRI-bound IgE-Fc (supplemental Fig. S3). Omalizumab inhibits not only the interaction between IgE-Fc and FcɛRI, but also the interaction between IgE-Fc and CD23 (30). Consistent with the latter, superposition on the Cɛ3 domains from the FabXol3/IgE-Fc structure, and the previously reported structure of CD23 in complex with an Fcɛ3–4 molecule (11), reveals steric clashes between FabXol3 and CD23 at both sites of CD23 engagement on the Cɛ3 domain.

    • Toll-like receptor 4 plays significant roles during allergic rhinitis

      2015, Allergologia et Immunopathologia
      Citation Excerpt :

      Allergic rhinitis, which is also known as nasal allergy and nasal hypersensitivity, is defined as an allergic condition of the nasal airways.6,7 It can be induced in individuals with a sensitised immune system via inhalation of an allergen, including pollen and dust or particles of shed skin and hair of animals (animal dander).8 In sensitive individuals, the allergen stimulates the production of IgE which binds to FCɛRI on mast cells and basophiles and induces secretion of several mediators including histamine, leukotrienes, prostaglandins and associated enzymes including tryptase.8

    • Asthma: Therapeutic update

      2014, Medicina Clinica
    View all citing articles on Scopus
    View full text